You are on page 1of 11

8594 Vol.

10, 8594 – 8602, December 15, 2004 Clinical Cancer Research

Epidermal Growth Factor Receptor (EGFR) Is Overexpressed in


Anaplastic Thyroid Cancer, and the EGFR Inhibitor Gefitinib
Inhibits the Growth of Anaplastic Thyroid Cancer

Bradley A. Schiff,2 Andrea B. McMurphy,2 slowed tumor growth in a nude mouse model of thyroid
Samar A. Jasser,1 Maher N. Younes,1 Dao Doan,1 carcinoma cells injected subcutaneously.
Conclusions: ATC cells consistently overexpress EGFR,
Orhan G. Yigitbasi,1 Seungwon Kim,1 Ge Zhou,1 rendering this receptor a potential target for molecular
Mahitosh Mandal,1 Benjamin N. Bekele,3 therapy. Gefitinib effectively blocks activation of EGFR by
F. Christopher Holsinger,1 Steven I. Sherman,4 EGF, inhibits ATC cellular proliferation, and induces apo-
Sai-Ching Yeung,5 Adel K. El-Naggar,1,6 and ptosis in vitro. Our in vivo results show that gefitinib has
Jeffrey N. Myers1,7 significant antitumor activity against ATC in a subcutane-
ous nude mouse tumor model and therefore is a potential
Departments of 1Head and Neck Surgery and 2Otorhinolaryngology,
Baylor College of Medicine, Houston, Texas; and Departments of candidate for human clinical trials.
3
Biostatistics, 4Endocrine Neoplasia and Hormonal Disorders,
5
Ambulatory Treatment and Emergency Care, 6Pathology, and INTRODUCTION
7
Cancer Biology, The University of Texas M. D. Anderson Cancer The annual prevalence of thyroid cancer in the United
Center, Houston, Texas
States was expected to be ⬃22,000 people in 2003 (1). The vast
majority of thyroid carcinomas are differentiated and can often
ABSTRACT be cured surgically. Anaplastic thyroid carcinomas are relatively
Purpose: No effective treatment options currently are rare, constituting only 1.6% all of the thyroid cancers (2), and
available to patients with anaplastic thyroid cancer (ATC), show extremely aggressive behavior, leading to a high mortality
rate. Patients with anaplastic thyroid cancer (ATC) face a uni-
resulting in high mortality rates. Epidermal growth factor
formly dismal prognosis, with average 5-year survival rates of
(EGF) has been shown to play a role in the pathogenesis of
⬍10% and a median survival time of 3 months (3). No effective
many types of cancer, and its receptor (EGFR) provides an
treatment options currently are available to patients with ATC.
attractive target for molecular therapy.
Targeted molecular therapy offers potential treatment al-
Experimental Design: The expression of EGFR was de-
ternatives for patients with ATC. Epidermal growth factor
termined in ATC in vitro and in vivo and in human tissue
(EGF) and its receptor (EGFR) have been implicated in the
arrays of ATC. We assessed the potential of the EGFR
pathogenesis of many different types of cancer and thus provide
inhibitor gefitinib (“Iressa,” ZD1839) to inhibit EGFR acti-
attractive targets for molecular therapy. EGFR, which is en-
vation in vitro and in vivo, inhibit ATC cellular proliferation, coded by the c-erb proto-oncogene, is a Mr 170,000 transmem-
induce apoptosis, and reduce the growth of ATC cells in vivo brane cell-surface glycoprotein consisting of an extracellular
when administered alone and in combination with pacli- ligand binding domain, a transmembrane domain, and an intra-
taxel. cellular domain with intrinsic tyrosine kinase activity (4, 5).
Results: EGFR was overexpressed in ATC cell lines in Dimerization of EGFR following the binding of the ligand
vitro and in vivo and in human ATC specimens. Activation of results in trans-phosphorylation of this receptor and subsequent
EGFR by EGF was blocked by the addition of gefitinib. In activation of several downstream signal transduction pathways,
vitro studies showed that gefitinib greatly inhibited cellular including the mitogen-activated protein kinase and phosphati-
proliferation and induced apoptosis in ATC cell lines and dylinositol-3⬘ kinase signaling pathways, which are involved in
promoting cellular proliferation and survival (6 – 8). In preclin-
ical studies, EGF has been shown to stimulate follicular cell
proliferation and to enhance the migration and invasiveness of
Received 4/8/04; revised 9/3/04; accepted 9/13/04.
papillary thyroid cancer (9 –11). The medical literature gener-
Grant support: The University of Texas M. D. Anderson Cancer ally supports the concept that EGFR, although expressed at
Center Physician-Scientist Program and Multi-Disciplinary Research higher levels in anaplastic and papillary cancers than in normal
Program in Thyroid Cancer and by The Golfers Against Cancer. Ge- thyroid tissue, is present in all of the thyroid tissues, but the data
fitinib was provided as a gift from AstraZeneca. in different studies often vary widely (9, 12–17). In an in vitro
The costs of publication of this article were defrayed in part by the
payment of page charges. This article must therefore be hereby marked study by Bergstrom et al. (18), EGFR was expressed in all six
advertisement in accordance with 18 U.S.C. Section 1734 solely to ATC cell lines examined, and constitutive phosphorylation of
indicate this fact. EGFR was found in three of the six cell lines tested.
Requests for reprints: Jeffrey N. Myers, Department of Head and Neck High expression of EGFR appears to be a negative prog-
Surgery, Unit 441, The University of Texas M. D. Anderson Cancer
Center, 1515 Holcombe Boulevard, Houston, TX 77030. Phone: 713-
nostic factor in multiple types of tumors, including breast cancer
745-2667; Fax: 713-794-4662; E-mail: jmyers@mdanderson.org. (19) and bladder cancer (20); however, few studies have exam-
©2004 American Association for Cancer Research. ined the clinical implications of EGFR expression and location

Downloaded from clincancerres.aacrjournals.org on April 14, 2021. © 2004 American Association for Cancer
Research.
Clinical Cancer Research 8595

in thyroid cancer. A recent study found a statistically significant L-glutamine, penicillin, sodium pyruvate, nonessential amino
correlation between the staining intensity of EGF and recurrence acids, and a twofold vitamin solution (Life Technologies, Inc.,
of papillary thyroid cancer (16). In another report, cytoplasmic Rockville, MD). Adherent monolayer cultures were maintained
immunopositivity was significantly associated with the extent of on plastic and incubated at 37°C in 5% carbon dioxide and 95%
primary tumor infiltration in papillary thyroid cancer, whereas air. The cultures were free of Mycoplasma species. All of the
membranous staining was not. Furthermore, in a multivariate cell lines injected into the mice tested free of the following
survival analysis, strong cytoplasmic EGFR staining of papillary pathogenic murine viruses: reovirus type 3, pneumonia virus, K
thyroid cancer was significantly associated with a decrease in virus, Theiler’s encephalitis virus, Sendai virus, minute virus,
recurrence-free survival (21). These findings suggest that the ectromelia virus, and lactate dehydrogenase virus (as assayed by
molecular blockage of EGFR activation has the potential to M.A. Bioproducts, Walkersville, MD). The cultures were main-
reduce thyroid tumor growth, making EGFR an attractive target tained no longer than 12 weeks after recovery from frozen
for molecular therapy against ATC. stocks.
Gefitinib (“Iressa,” ZD1839; AstraZeneca, Wilmington, Reagents. For in vitro administration, gefitinib was dis-
DE), a synthetic anilinoquinazoline, is an orally active EGFR solved in dimethyl sulfoxide to a concentration of 20 mmol/L.
inhibitor that is highly selective, with minimal activity against For in vivo testing, gefitinib was dissolved in a lactate salt
other tyrosine kinases. Gefitinib has been shown to block EGF- solution. Propidium iodide (PI) and tetrazolium (MTT) were
stimulated EGFR autophosphorylation (22) and EGFR-medi- purchased from Sigma-Aldrich Corp. (St. Louis, MO).
ated downstream signal transduction. The drug also has shown Western Immunoblot Analysis. All of the cells were
activity against a variety of human tumor cells in vitro and incubated in serum-free medium for 24 hours. Cells were incu-
additive to synergistic effects when combined with radiation bated with gefitinib for 1 hour at concentrations ranging from
therapy or chemotherapy (23, 24). Results of Phase I trials have 0.01 to 100 ␮mol/L before addition of EGF (40 ng/mL) for 15
indicated that this agent offers good bioavailability and tolera- minutes. The cells then were washed with PBS, and lysis buffer
bility (25). Many Phase II and III studies currently are underway was added [1% Triton X-100, 20 mmol/L Tris (pH 8.0), 137
to assess the effectiveness of gefitinib against a variety of mmol/L sodium chloride, 10% glycerol (v/v), 2 mmol/L EDTA,
carcinomas, including breast, lung, colorectal, head and neck, 1 mmol/L phenylmethylsulfonyl fluoride, 20 ␮mol/L aprotinin-
prostate, and renal. Two recent studies have suggested that leupeptin-trypsin inhibitor, and 2 mmol/L sodium orthovana-
response to gefitinib therapy is linked to mutations in the EGFR
date]. The cells were scraped and spun in a centrifuge to remove
gene (26, 27). Although numerous studies already have shown
insoluble proteins. The samples were diluted in sample buffer
that gefitinib possesses clinically meaningful antitumor activity
[10% SDS, 0.5 mmol/L Tris-HCl (pH 6.8), 1 mol/L dithiothre-
in several malignancies (28), to our knowledge, no clinical trials
itol, 10% (v/v) glycerol, and 1% bromphenol blue] and boiled.
yet exist to determine the effectiveness of gefitinib against ATC.
The proteins (50 ␮g) were resolved on polyacrylamide gel
Thus, we investigated the role of EGFR and its inhibitor ge-
electrophoresis and electrophoretically transferred onto 0.45-␮g
fitinib in ATC to determine whether gefitinib possesses mean-
nitrocellulose membranes. The membranes were blocked with
ingful antitumor activity against ATC, potentially justifying
5% (w/v) nonfat milk in 0.1% Tween 20 (v/v) in Tris-buffered
clinical trials.
saline, probed with rabbit monoclonal anti-EGFR (1:2000; Up-
state Biotechnology, Inc., Lake Placid, NY) in 1% nonfat milk,
MATERIALS AND METHODS and incubated with peroxidase-conjugated sheep antirabbit im-
Animals. Male athymic nude mice, ages 8 to 12 weeks, munoglobulin (1:2000; Amersham, Piscataway, NJ) in 1% non-
were purchased from the animal production area of the National fat milk. The blots also were probed with rabbit anti–phospho-
Cancer Institute Frederick Cancer Research and Development EGFR (p-EGFR 1068; Cell Signaling Technologies, Beverly,
Center (Frederick, MD). The mice were housed and maintained MA), diluted 1:1000 in 1% nonfat milk, and incubated with
in laminar flow cabinets under specific pathogen-free conditions peroxidase-conjugated sheep antirabbit IgG (1:3000). All of the
in facilities approved by the American Association for Accred- blots were probed with anti–␤-actin (1:1000) in 1% nonfat milk,
itation of Laboratory Animal Care in accordance with current followed by horseradish peroxidase– conjugated donkey antirab-
regulations and standards of the United States Department of bit IgG (1:2000; Amersham) in 1% nonfat milk. Protein bands
Agriculture, the United States Department of Health and Human were visualized using an enhanced chemiluminescence detec-
Services, and the NIH. The mice were treated in accordance tion system (Amersham).
with the Animal Care and Use Guidelines of the University of ELISA. Cells from the anaplastic cancer cell lines
Texas M. D. Anderson Cancer Center (Houston, TX) under a KAT-4, K18, C643, HTH, ARO, and DRO and from the pap-
protocol approved by the Institutional Animal Care Use Com- illary cancer cell line NPA187 were grown in serum-free me-
mittee. dium and treated with gefitinib at concentrations ranging from
Cell Lines and Culture Conditions. The papillary thy- 0.01 to 100 ␮mol/L. After 24 hours, the supernatant was col-
roid carcinoma cell line NPA187 and the ATC cell lines KAT-4, lected, and the cells were counted. The supernatants were ex-
K18, C643, HTH, ARO, and DRO were used. Oral cavity amined for transforming growth factor ␣ (TGF-␣) and EGF. All
squamous cell cancer line TU167 was used as a positive control, of the kits were purchased from R&D Systems (Minneapolis,
and the fibroblast line 3T3 was used as a negative control. The MN), and all of the experiments were performed according to
cells were grown in Dulbecco’s modified Eagle’s medium sup- the manufacturer’s instructions. The absorbance and concentra-
plemented with 10% fetal bovine serum (DMEM-10% FBS), tion (pg/mL) were measured using a microplate reader at 450

Downloaded from clincancerres.aacrjournals.org on April 14, 2021. © 2004 American Association for Cancer
Research.
8596 EGFR Inhibitor Gefitinib Inhibits ATC

nm. For each cell line, the results were plotted as the ratio of the kon Microphot-FXA (Nikon Inc., Tokyo, Japan) equipped with
concentration to the total number of cells. an HBO 100 mercury lamp and narrow bandpass filters to
PCR Analysis of EGFR Mutations. The PCR was used individually select for green, red, and blue fluorescence
to amplify exons 18, 19, and 21 from the EGFR gene using (Chroma Technology Corp., Brattleboro, VT). Images were
genomic DNA isolated from the following tumor-derived cell captured using a cooled charge-coupled device Hamamatsu
lines: papillary thyroid cancer cell line NPA187 and ATC cell 5810 camera (Hamamatsu Corp., Bridgewater, NJ) and Optimas
lines DRO, K18, ARO, HTH-74, C643, and KAT-4. PCR am- Image Analysis software (Media Cybernetics, Silver Spring,
plicons were purified using QIAquick PCR purification kit MD). Photomontages were prepared using Adobe Photoshop
(Qiagen, Valencia CA) and then sent to LoneStar Labs (Hous- software (Adobe Systems Inc., San Jose, CA).
ton, TX) for sequencing. Electropherograms were analyzed for Human Thyroid Tissue Arrays. Thyroid tumor tissue
the presence of mutations. arrays representative of the entire spectrum of benign and ma-
Measurement of Cell Proliferation. Gefitinib was lignant neoplasms, including ATC constructed at the head and
tested on the KAT-4 ATC cell line using an MTT-based assay, neck tissue care facility, were used to screen for EGFR expres-
which measures cell proliferation based on the ability of live sion. The arrays represented 14 papillary carcinomas, 6 anaplas-
cells to convert MTT to dark blue formazan. Two thousand cells tic carcinomas, and 9 samples of nondiseased thyroid tissue.
were grown in DMEM-10% FBS, with and without 0.01 to 100 Two cores of each sample were placed differentially in the
␮mol/L gefitinib, in 96-well tissue culture plates. After 24 recipient block. Two pathologists scored these blindly and in-
hours, the cells again were incubated with medium alone or dependently on a scale of 0 to 3. Whenever a discrepancy in
medium containing various concentrations of gefitinib diluted in scoring was noted, both pathologists reexamined the sample in
dimethyl sulfoxide. After a 3-day incubation, the number of
question, and a consensus was reached.
metabolically active cells was determined by MTT assay using
In vivo Xenografts. Tumor response to gefitinib was
a 96-well microtiter plate reader (MR-5000; Dynatech Labora-
gauged using a nude mouse model of thyroid cancer. KAT-4
tories Inc., Chantilly, VA) at an absorbance of 570 nm. The
ATC cells were harvested from subconfluent cultures by
absorbance for a control plate, which was not seeded with any
trypsinization and then washed. Using a 30-gauge needle under
cells during initial plating, was subtracted from the absorbance
direct visualization, 5 ⫻ 106 KAT-4 cells diluted in 30 ␮L of
of every other well.
serum-free medium were injected subcutaneously into the cer-
Measurement of Cell Death. Cells were plated at a
density of 5 ⫻ 105 cells in 38 mm2 six-well plates (Costar, vical area. Tumors were allowed to grow for 14 days; all of the
Cambridge, MA) and maintained for 24 hours before treatment mice then were weighed, and all of the tumors were measured
with gefitinib. After 24 hours, gefitinib was added at various using microcalipers.
concentrations. After treatment for 48 hours, PI staining of Tumor volume was calculated using the formula
hypodiploid DNA determined the extent of cell death. A total of (A)(B2)␲/6, where A was the length of the longest aspect of the
3 ⫻ 105 cells were resuspended in a Nicoletti buffer [50 ␮g/mL tumor, and B was the length of the tumor perpendicular to A.
PI, 0.1% sodium citrate, 0.1% Triton X-100, and 1 mg/mL The two mice with the largest tumors and the three with the
RNase (Roche, Basel, Switzerland) in PBS] for 20 minutes at smallest tumors were excluded from the analysis. All of the
4°C. Cells then were analyzed by flow cytometry, and the other mice then were placed into groups of five mice with
sub-G0/G1 fraction was measured using the Lysys software similar average tumor volumes. The groups then were random-
(Becton Dickinson, Franklin Lakes, NJ). ized into seven treatment groups. Each mouse in group 1, the
Immunohistochemical Analysis of Murine Tumor Fro- control group, received sodium lactate at a dosage of 0.2 mL/d
zen Sections and Human Tumor Tissue Arrays. Immuno- for 5 days (Monday through Friday) for 2 weeks via oral gavage.
histochemical analysis was performed using rabbit anti-EGFR All of the other groups received gefitinib dissolved in 0.2 mL of
antibodies (Santa Cruz Biotechnology, Santa Cruz, CA). Frozen sodium lactate solution via oral gavage. The mice in group 2
tumors were sectioned (8 to 10 ␮m thick), mounted on posi- each received 0.9 mg/d (30 mg/kg) for 5 days per week for 2
tively charged Superfrost slides (Fisher Scientific, Houston, weeks; mice in group 3, 1.8 mg/d (60 mg/kg) for 5 days per
TX), air dried for 30 minutes, and fixed in cold acetone for 10 week for 2 weeks; mice in group 4, 2.7 mg/d (90 mg/kg) for 5
minutes. The slides were washed three times with PBS (pH 7.5), days per week for 2 weeks; mice in group 5, 4.5 mg/d (150
blocked for 20 minutes at room temperature in PBS supple- mg/kg) for 5 days per week for 2 weeks; mice in group 6, 4.5
mented with 1% normal goat serum and 5% normal horse serum mg/d (150 mg/kg) for 5 days per week for 2 weeks ⫹ paclitaxel
(protein-blocking solution), and incubated with primary anti- (200 ␮g/wk injected intraperitoneally); and mice in group 7, 4.5
body for 18 hours at 4°C. Samples then were washed three times mg/d (150 mg/kg) every other day (QOD), Monday, Wednes-
for 3 minutes, blocked with protein-blocking solution for 10 day, and Friday for 2 weeks. The mice were weighed and the
minutes, and incubated with AlexaFluor 594 – conjugated goat tumors were measured on days 8 and 12 using microcalipers
antirabbit IgG (Molecular Probes, Eugene, OR) for 1 hour at until the mice were euthanatized after 2 weeks of treatment.
room temperature in the dark. Samples again were washed three After the mice were euthanatized, the tumors again were meas-
times for 5 minutes and then counterstained with 300 ␮g/mL ured, and the mice were weighed.
Hoechst stain for 1 to 2 minutes at room temperature. The slides For immunohistochemical and routine hematoxylin and
were washed again with PBS and then mounted using propyl eosin staining, one part of the tissue was fixed in formalin and
gallate. embedded in paraffin, and another part was embedded in opti-
Immunofluorescence microscopy was performed in a Ni- mal cutting-temperature compound (Miles Inc., Elkhart, IN),

Downloaded from clincancerres.aacrjournals.org on April 14, 2021. © 2004 American Association for Cancer
Research.
Clinical Cancer Research 8597

Table 1 Level of staining for EGFR in normal, papillary, and RESULTS


anaplastic thyroid tissue from 29 human subjects
Production of Growth Factors EGF and TGF-␣ by
EGFR staining Thyroid Cancer Cell Lines. To assess whether ATC cells
Level of staining Percentage produce the EGFR ligands EGF and TGF-␣, ELISA was used to
Tissue No. of of specimen assess their levels in culture supernatants from six anaplastic
type subjects 0 1 2 3 staining and one papillary cancer cell lines. EGF was not present at an
Normal 9 9 0 0 0 0 appreciable level in the supernatants from any of the cell lines
Papillary 14 12 2 0 0 14 examined. TGF-␣ was present at a level of 38.5 pg/mL per cell
Anaplastic 6 1 3 2 0 83
number in the medium from the ATC cell line DRO but was not
Abbreviation: EGFR, epidermal growth factor receptor. present at an appreciable level in any of the other cell lines
examined.
Expression of EGFR in Human Tissue Arrays. To
determine the expression level of EGFR in normal and neoplas-
rapidly frozen in liquid nitrogen, and stored at ⫺80°C. Hema-
tic human thyroid tissue, tissue arrays of surgical specimens
toxylin and eosin staining confirmed the presence of tumors.
composed of normal thyroid tissue, papillary thyroid cancer, and
Statistical Analyses. To examine the results of EGFR in
ATC were obtained from Dr. Adel El-Naggar (Department of
human thyroid tissue arrays, we performed three pairwise com-
Surgical Pathology, M. D. Anderson Cancer Center). None of
parisons using Fisher’s exact test. In the first analysis, the type
I error rate was controlled at 0.017, guaranteeing that the overall the nine normal tissue specimens stained positive for EGFR. Of
type I error rate would be controlled at 0.05. To determine the the specimens from the 14 patients with papillary thyroid can-
IC50 for MTT and apoptosis assays, linear interpolation was cer, 2 stained positive for EGFR. Specimens from five of the six
used. We chose this method because the changes in absorbance patients with ATC stained positive for EGFR; specimens from
as a percentage of the control and in apoptosis were linearly two of these five patients stained at level 2, whereas specimens
related to gefitinib levels in the range of the IC50. from the three other patients stained at level 1. Comparisons of
Two methods were used to analyze the data from the study ATC specimens with normal thyroid tissue specimens and ATC
involving the model in which nude mice were injected subcu- specimens with papillary thyroid cancer specimens revealed
taneously. The purpose of this analysis was to determine statistically significant differences in EGFR expression (P ⫽
whether there was a statistically significant difference in change 0.002 and P ⫽ 0.007, respectively). No statistically significant
in tumor size between mice treated with gefitinib at dosages of difference in EGFR expression was found between the papillary
30, 60, 90, and 150 mg/d, 150 mg/d plus paclitaxel, and the thyroid cancer and the normal thyroid tissue specimens (P ⫽
control group. The endpoint of interest was the percentage 0.5; Table 1, Fig. 1).
change from initial tumor size at day 12 of treatment. The Expression of EGFR/Phospho-EGFR in Human Thy-
Wilcoxon rank sum test was used to detect statistical signifi- roid Cancer Cell Lines. To assess expression of EGFR and
cance in the percentage change in tumor size from the initial p-EGFR in human thyroid tissue cell lines in vitro, Western blot
tumor size at day 12. The Jonckheere-Terpstra test was used to analysis was performed on cellular lysates of the six ATC cell
test for evidence of a trend of decreasing tumor size with lines (ARO, C643, DRO, HTH, K18, and KAT-4) and the
increasing treatment dosage. papillary cell line (NPA187). Five of the six ATC cell lines (all

Fig. 1 Representative samples of the tissue arrays for normal thyroid tissue, papillary thyroid cancer, and ATC after staining for EGFR. Staining
levels were 0, 1, and 2, respectively.

Downloaded from clincancerres.aacrjournals.org on April 14, 2021. © 2004 American Association for Cancer
Research.
8598 EGFR Inhibitor Gefitinib Inhibits ATC

but did not express p-EGFR if euthanatized 6 hours after the


final treatment. The mice treated with gefitinib at a dosage of
150 mg/kg/d showed no expression of p-EGFR, whether eu-
thanatized either immediately before or 6 hours after the final
treatment (Fig. 4).
Gefitinib Inhibited Cellular Proliferation of KAT-4
Cells In vitro. To determine the effect of inhibition of EGFR
signaling on ATC cell proliferation in vitro, three MTT assays
Fig. 2 Western blot analysis of cellular lysates of six ATC cell lines were performed using gefitinib at concentrations ranging from 1
(ARO, C643, DRO, K18, and KAT-4) and the papillary cell line to 20 ␮mol/L (Fig. 5). The lowest concentration at which
NPA187. Five of the six ATC cell lines (all except DRO) stained cellular proliferation was inhibited was 6 ␮mol/L. The IC50 was
positive for EGFR, whereas NPA187 was negative for EGFR. 8.36 ␮mol/L, and maximal inhibition occurred at a concentra-
tion of 14 ␮mol/L.
Gefitinib Induced Apoptosis in KAT-4 Cells In vitro.
To assess the effects of gefitinib on induction of cell death of
except DRO) stained positive for EGFR (Fig. 2), and the pap- ATC cells, we performed a PI apoptosis assay on KAT-4 cells
illary cell line, NPA187, was negative for EGFR. In additional after 48 hours of treatment. When treated at a concentration of
Western blot analyses, all of the cell lines were negative for the 8 ␮mol/L (the IC50 for the MTT assays), 5.5% of the cells
activated, or phosphorylated, form of EGFR (p-EGFR) unless underwent apoptosis. At a concentration of 12 ␮mol/L, 27.6%
stimulated with EGF (data not shown). of the cells were apoptotic; at 19 ␮mol/L, 54.7%; at 22 ␮mol/L,
In vivo Expression of EGF, EGFR, and p-EGFR. A 80.7%; and at 50 ␮mol/L, 94.5% (Fig. 6). The estimated IC50
nude mouse model was used to determine the expression of for the apoptosis assays was 18.35 ␮mol/L.
EGF, EGFR, and p-EGFR in mice implanted subcutaneously Gefitinib Alone and in Combination with Paclitaxel
with 2.5 ⫻ 106 cells of KAT-4 ATC. Tumors were allowed to Reduced KAT-4 Tumor Size and Growth in a Nude Mouse
form, and after 1 month, the mice were euthanatized, and the Model of Thyroid Cancer. A nude mouse model of thyroid
tumors were subjected to immunohistochemical staining. Nor- cancer was used for the in vivo portion of the study to assess the
mal murine thyroid tissue and ATC implants expressed EGFR in vivo antitumor activity of gefitinib. At day 12, the control
and p-EGFR. group of mice showed an increased tumor size of 173% of the
PCR Analysis of EGFR Showed No Mutations. Anal- initial tumor size. The groups treated with gefitinib at dosages of
ysis of exons 18, 19, and 21 of EGFR showed no mutations in 30, 60, 90, and 150 mg/kg/d had increased tumor sizes at day 12
the six ATC cell lines tested (DRO, K18, ARO, HTH-74, C643, of 158%, 165%, 112%, and 104% of the initial tumor size,
and KAT-4) or the papillary cell line NPA187. respectively. The group treated with paclitaxel alone had an
Gefitinib Inhibited EGFR Phosphorylation in KAT-4 increase in tumor size at day 12 of 134% of initial tumor size.
Cells In vitro. To assess whether gefitinib could inhibit EGFR In the group treated with 150 mg/kg/d plus paclitaxel, tumors
signaling in ATC in vivo, KAT-4 ATC cell lines were exposed decreased to 98% of initial tumor size (Fig. 7).
to serum-free medium for 24 hours. The cells then were treated
with various concentrations of gefitinib in serum-free medium DISCUSSION
for 1 hour. The level of gefitinib ranged from 0.01 to 100 Because patients with ATC face a dismal prognosis and
␮mol/L. After 1 hour of exposure, recombinant EGF at a dosage have no effective treatment options, novel treatments are
of 40 ng/mL was added for 15 minutes. In Western blot analysis, needed. The EGFR is involved in the pathogenesis or progres-
the KAT-4 cells showed a high level of p-EGFR when stimu- sion of many different types of cancers and therefore is a
lated with EGF. Gefitinib partially blocked EGF autophospho- potential target for molecular therapy in ATC. However, little
rylation of EGFR at a concentration of 0.01 ␮mol/L and almost
completely blocked autophosphorylation at 1 ␮mol/L. At a
concentration of 10 ␮mol/L, gefitinib completely blocked
EGFR autophosphorylation (Fig. 3).
Gefitinib Inhibited EGFR Phosphorylation in KAT-4
Cells In vivo. To assess the efficacy of gefitinib inhibition of
autophosphorylation of EGFR in ATC cells in vivo, mice im-
planted subcutaneously with the KAT-4 ATC cell line, which
expresses a high level of p-EGFR, were treated with gefitinib at
dosages of 30, 60, 90, and 150 mg/kg/d and at a dosage of 150
mg/kg/QOD. The mice treated with gefitinib at 30 and 60
mg/kg/d showed high levels of p-EGFR. The tumor sections
from the mice treated with gefitinib at 90 mg/kg/d showed high Fig. 3 Western blot analysis of KAT-4 ATC cell lines treated with
levels of p-EGFR but lower expression than in tumor sections various concentrations of gefitinib and subsequently exposed to EGF.
Gefitinib partially blocked EGF autophosphorylation of EGFR at a
from the mice treated with 30 or 60 mg/kg/d. The mice treated concentration of 0.01 ␮mol/L and almost completely blocked autophos-
with gefitinib at 150 mg/kg/QOD showed p-EGFR expression if phorylation at 1 ␮mol/L. At a concentration of 10 ␮mol/L, gefitinib
they were euthanatized immediately before the final treatment completely blocked EGFR autophosphorylation.

Downloaded from clincancerres.aacrjournals.org on April 14, 2021. © 2004 American Association for Cancer
Research.
Clinical Cancer Research 8599

Fig. 4 Immunohistochemical results of mice subcutaneously implanted with KAT-4 ATC cell line and treated with various dosages of gefitinib. The
tumors of mice treated with 30 and 60 mg/kg QD (QD, every day) of gefitinib showed high levels of p-EGFR, whereas the tumors of mice treated
with 90 mg/kg/d showed lower expression of p-EGFR. The tumors of mice treated with 150 mg/kg QOD (QOD, every other day) showed p-EGFR
expression if the mice were euthanatized immediately before the final treatment with gefitinib (48 hours after the previous gefitinib treatment) but
did not express p-EGFR if sacrificed 6 hours after the final treatment. The tumors of mice treated with 150 mg/kg/d of gefitinib showed no expression
of p-EGFR whether the mice were euthanatized either immediately before or 6 hours after the final treatment with gefitinib. This showed that gefitinib
at a dosage of 150 mg/kg/d can completely block expression of p-EGFR in a nude mouse model of thyroid carcinoma. * Killed immediately before
treatment. ** Killed 6 hours after treatment.

research has been done to examine the role of EGFR in ATC


and the effectiveness of anti-EGFR therapies against ATC.
Gefitinib is an EGFR tyrosine kinase inhibitor that has already
been shown to have a favorable safety and tolerability profile in
numerous Phase I clinical trials and therefore is a promising area
of investigation in the search for effective treatments for patients
with ATC.
To initiate studies, we performed ELISA to determine the
expression of growth factors TGF-␣ and EGF. EGF was not
present in any of the six ATC cell lines examined, and TGF-␣
was present only in the DRO cell line. DRO, the only ATC cell
line that did not express EGFR, also was the only tested cell line
that expressed TGF-␣. In papillary thyroid cancer, TGF-␣ and
EGFR expression often are linked.
Our thyroid tissue arrays showed significantly greater
EGFR expression in ATC specimens than in either normal
thyroid tissue or papillary thyroid cancer specimens (P ⫽ 0.002
and 0.007, respectively). The medical literature on this topic is
mixed. Some studies (29) report that EGFR is expressed only in
diseased thyroid tissue, whereas others report that EGFR is
expressed in all of the thyroid tissues (9). However, most studies
suggest that EGFR is expressed at a higher level in thyroid
cancer than in normal thyroid tissue (30, 31). The data from our
tissue arrays support this finding and suggest that EGFR is
highly expressed in ATC. Fig. 5 MTT assay of KAT-4 ATC cells exposed to various concentra-
tions of gefitinib. The lowest concentration at which cellular prolifera-
Our analysis of the expression of EGFR by Western blot
tion was inhibited was 6 ␮mol/L, and the IC50 was 8.36 ␮mol/L.
analysis showed no expression of EGFR in the papillary cell line Maximal inhibition occurred at a concentration of 14 ␮mol/L. Thus,
NPA187 but did show EGFR expression in five of the six ATC gefitinib is able to effectively inhibit ATC cellular proliferation.

Downloaded from clincancerres.aacrjournals.org on April 14, 2021. © 2004 American Association for Cancer
Research.
8600 EGFR Inhibitor Gefitinib Inhibits ATC

Fig. 6 PI apoptosis assay on KAT-4 cells after 48


hours of treatment with gefitinib. When treated at
a concentration of 8 ␮mol/L (the IC50 for the MTT
assays), 5.5% of the cells underwent apoptosis.
The estimated IC50 for the apoptosis assays was
18.4 ␮mol/L. At a gefitinib concentration of 22
␮mol/L, 80.7% of cells underwent apoptosis,
whereas at 50 ␮mol/L, 94.5% of cells underwent
apoptosis. This shows that gefitinib is able to
induce apoptosis in ATC cell lines.

cell lines. The results also showed that EGFR was not consti- We also showed that gefitinib blocked EGF-mediated ac-
tutively phosphorylated in any of the ATC cell lines tested, but tivation of EGFR on ATC cell lines in vitro. The mice treated
EGFR phosphorylation was readily observed in these cell lines with 30 and 60 mg/kg/d of gefitinib showed high levels of
after stimulation with EGF. These data are in concordance with p-EGFR. Specimens from the mice treated with 90 mg/kg/d of
those from previous studies that suggest that EGFR is expressed gefitinib showed high levels of p-EGFR but did not stain as
in ATC cell lines, but they contradict the literature that suggests positively as did those from the mice treated with 30 or 60
that EGFR is constitutively activated in some ATC cell lines. mg/kg/d. The mice treated with gefitinib 150 mg/kg/d showed
However, immunohistochemical staining of sections of subcu- no expression of p-EGFR regardless of whether they were
taneously implanted KAT-4 tumors revealed that those tumors euthanatized 6 hours after the final treatment or immediately
constitutively expressed EGFR and p-EGFR, as did normal before the final treatment was scheduled, a fact that indicates
murine thyroid tissue, suggesting that EGFR activation is up- that gefitinib at a dosage of 150 mg/kg/d is able to continuously
regulated in vivo. suppress EGFR phosphorylation. The mice treated with 150

Fig. 7 A nude mouse model of thyroid


carcinoma cells injected subcutaneously
was used to assess the in vivo antitumor
activity of gefitinib. At day 12, the control
group of mice showed an increased tumor
size of 173% of the initial tumor size. The
groups treated with gefitinib at dosages of
30, 60, 90, and 150 mg/kg/d had increased
tumor sizes at day 12 of 158%, 165%,
112%, and 104% of the initial tumor size,
respectively. The group treated with pacli-
taxel alone had an increase in tumor size at
day 12 of 134% of initial tumor size. In the
group treated daily with 150 mg/kg/d ge-
fitinib ⫹ paclitaxel, tumors decreased to
98% of initial tumor size. This shows that
gefitinib, alone and in combination with
paclitaxel, was able to reduce the growth
of ATC in a nude mouse model of thyroid
carcinoma cells injected subcutaneously.

Downloaded from clincancerres.aacrjournals.org on April 14, 2021. © 2004 American Association for Cancer
Research.
Clinical Cancer Research 8601

mg/kg/QOD of gefitinib showed p-EGFR expression if euthana- 5. Carpenter G, Cohen S. Epidermal growth factor. J Biol Chem 1990;
tized immediately before the final treatment was supposed to be 265:7709 –12.
given but did not express p-EGFR if euthanatized 6 hours after 6. Ullrich A, Schlessinger J. Signal transduction by receptors with
the final treatment, showing that a dosage of 150 mg/kg is tyrosine kinase activity. Cell 1990;61:203–12.
unable to suppress EGFR phosphorylation for 48 hours. Our 7. Aaronson SA. Growth factors and cancer. Science 1991;254:
1146 –53.
data show that gefitinib at a daily dosage of 150 mg/kg is able
8. Marti U, Ruchti C, Kampf J, et al. Nuclear localization of epidermal
to suppress EGFR activation for 24 to 48 hours in a nude mouse
growth factor and epidermal growth factor receptors in human thyroid
model of thyroid cancer. tissues. Thyroid 2001;11:137– 45.
After establishing that EGFR is overexpressed in ATC and 9. van der Laan BF, Freeman JL, Asa SL. Expression of growth factors
that gefitinib can suppress EGFR phosphorylation in vitro and in and growth factor receptors in normal and tumorous human thyroid
vivo, we then examined the effect of gefitinib on ATC cell tissues. Thyroid 1995;5:67–73.
growth and death. An MTT assay showed that 12 ␮mol/L of 10. Hoelting T, Siperstein AE, Clark OH, Duh QY. Epidermal growth
gefitinib caused near-total growth inhibition. A PI apoptosis factor enhances proliferation, migration, and invasion of follicular and
assay revealed that 22 ␮mol/L of gefitinib induced a rate of papillary thyroid cancer in vitro and in vivo. J Clin Endocrinol Metab
1994;79:401– 8.
apoptosis ⬎80%. Thus, gefitinib is able to effectively inhibit
11. Bechtner G, Schopohl D, Rafferzeder M, Gartner R, Welsch U.
ATC cellular proliferation and induce apoptosis in ATC cell
Stimulation of thyroid cell proliferation by epidermal growth factor is
lines, providing further evidence that gefitinib is effective different from cell growth induced by thyrotropin or insulin-like growth
against ATC. factor I. Eur J Endocrinol 1996;134:639 – 48.
The final portion of our experiments used a nude mouse 12. Lemoine NR, Hughes CM, Gullick WJ, Brown CL, Wynford-
model to examine the effects of various dosing schedules of Thomas D. Abnormalities of the EGF receptor system in human thyroid
gefitinib alone or in combination with paclitaxel on subcutane- neoplasia. Int J Cancer 1991;49:558 – 61.
ously implanted KAT-4 tumors. The difference in change in 13. Westermark K, Lundqvist M, Wallin G, et al. EGF-receptors in
tumor size between the control group and the group treated with human normal and pathological thyroid tissue. Histopathology 1996;28:
221–7.
gefitinib at a dosage of 150 mg/d ⫹ paclitaxel approached but
did not achieve statistical significance (P ⫽ 0.06). However, if 14. Duh QY, Gum ET, Gerend PL, Raper SE, Clark OH. Epidermal
growth factor receptors in normal and neoplastic thyroid tissue. Surgery
a trend analysis is done looking at increasing doses of gefitinib 1985;98:1000 –7.
(and including gefitinib 150 mg/d ⫹ paclitaxel as the group
15. Duh QY, Siperstein AE, Miller RA, Sancho JJ, Demeure MJ, Clark
receiving the highest level of treatment), there is a significant OH. Epidermal growth factor receptors and adenylate cyclase activity in
difference between change in tumor size and increasing treat- human thyroid tissues. World J Surg 1990;4:410 – 8.
ment dosage (P ⫽ 0.015). 16. Mizukami Y, Nonomura A, Hashimoto T, et al. Immunohistochem-
In summary, EGF has been shown to play a role in the ical demonstration of epidermal growth factor and c-myc oncogene
pathogenesis of many types of cancer, and its receptor provides product in normal, benign and malignant thyroid tissues. Histopathology
a promising target for molecular therapy. Little research has 1991;18:11– 8.
been done to examine the role of EGFR in ATC. Novel treat- 17. Aasland R, Akslen LA, Varhaug JE, Lillehaug JR. Co-expression of
ment strategies are needed for patients with ATC because cur- the genes encoding transforming growth factor-␣ and its receptor in
papillary carcinomas of the thyroid. Int J Cancer 1990;46:382–7.
rent treatment options offer little benefit. We showed that EGFR
18. Bergström JD, Westermark B, Heldin N-E. Epidermal growth factor
is increased in ATC cell lines in vitro and in vivo and in human
receptor signaling activates Met in human anaplastic thyroid carcinoma
ATCs. Gefitinib was able to inhibit EGFR phosphorylation in cells. Exp Cell Res 2000;259:293–9.
vitro and in vivo and to reduce cellular proliferation and induce 19. Nicholson S, Richard J, Sainsbury C, et al. Epidermal growth factor
apoptosis in ATC cell lines. Finally, gefitinib alone and in receptor (EGFr); results of a 6 year follow-up study in operable breast
combination with paclitaxel was able to reduce the growth of cancer with emphasis on the node negative subgroup. Br J Cancer
ATC cells in a nude mouse model of thyroid carcinoma cells 1991;63:146 –50.
injected subcutaneously. The favorable safety profile of ge- 20. Lipponen P, Eskelinen M. Expression of epidermal growth factor
fitinib has already been proven in Phase I clinical trials, and receptor in bladder cancer as related to established prognostic factors,
based on the data presented here, clinical trials of gefitinib in oncoprotein (c-erbB-2, p53) expression and long-term prognosis. Br J
Cancer 1994;69:1120 –5.
patients with ATC are warranted.
21. Akslen LA, Myking AO, Salvesen H, Varhaug JE. Prognostic
impact of EGF-receptor in papillary thyroid carcinoma. Br J Cancer
1993;68:808 –12.
REFERENCES
22. Wakeling AE, Guy SP, Woodburn JR, et al. ZD1839 (Iressa): an
1. Jemal A, Murray T, Samuels A, Ghafoor A, Ward E, Thun MJ.
orally active inhibitor of epidermal growth factor signaling with poten-
Cancer statistics, 2003. CA Cancer J Clin 2003;53:5–26.
tial for cancer therapy. Cancer Res 2202;62:5749 –54.
2. Gilliland FD, Hunt WC, Morris DM, Key CR. Prognostic factors for
thyroid carcinoma. A population-based study of 15,698 cases from the 23. Ciardiello F. EGFR-targeted agents potentiate the antitumour activ-
Surveillance, Epidemiology and End Results (SEER) program 1973– ity of chemotherapy and radiotherapy. Signal 2002;2:4 –11.
1991. Cancer 1997;79:564 –73. 24. Sirotnak FM, Zakowski MF, Miller VA, Scher HI, Kris MG.
3. McIver B, Hay ID, Giuffrida DF, et al. Anaplastic thyroid carcinoma: Efficacy of cytotoxic agents against human tumor xenographs is
a 50-year experience at a single institution. Surgery 2001;130:1028 –34. markedly enhanced by coadministration of ZD1839 (Iressa), an inhibitor
4. Ullrich A, Coussens L, Hayflick JS, et al. Human epidermal growth of EGFR tyrosine kinase. Clin Cancer Res 2000;6:4885–92.
factor receptor cDNA sequence and aberrant expression of the amplified 25. Ranson M. ZD1839 (Iressa): for more than just non-small cell lung
gene in A431 epidermoid carcinoma cells. Nature 1984;309:418 –25. cancer. Oncologist 2002;7(Suppl 4):16 –24.

Downloaded from clincancerres.aacrjournals.org on April 14, 2021. © 2004 American Association for Cancer
Research.
8602 EGFR Inhibitor Gefitinib Inhibits ATC

26. Lynch TJ, Bell DW, Sordella R, et al. Activating mutations in 29. Haugen DR, Akslen LA, Varhaug JE, Lillehaug JR. Demonstration
the epidermal growth factor receptor underlying responsiveness of of a TGF-␣-EGF-receptor autocrine loop and c-myc protein over-
non-small-cell lung cancer to gefitinib. N Engl J Med 2004;350: expression in papillary thyroid carcinomas. Int J Cancer 1993;55:37– 43.
2129 –39. 30. Aasland R, Akslen LA, Varhaug JE, Lillehaug JR. Co-expression of
27. Paez JG, Jänne PA, Lee JC, et al. EGFR mutations in lung cancer: the genes encoding transforming growth factor-␣ and its receptor in
correlation with clinical response to gefitinib therapy. Science 2004; papillary carcinomas of the thyroid. Int J Cancer 1990;46:382–7.
304:1497–500. 31. Gorgoulis V, Aninos D, Priftis C, et al. Expression of epidermal
28. Herbst RS. ZD1839: targeting the epidermal growth factor receptor growth factor, transforming growth factor-␣ and epidermal growth
in cancer therapy. Expert Opin Investig Drugs 2002;11:837– 49. factor receptor in thyroid tumors. In Vivo 1992;6:291– 6.

Downloaded from clincancerres.aacrjournals.org on April 14, 2021. © 2004 American Association for Cancer
Research.
Editor's Note Clinical
Cancer
Research
Editor's Note: Epidermal Growth Factor
Receptor (EGFR) Is Overexpressed in
Anaplastic Thyroid Cancer, and the EGFR
Inhibitor Gefitinib Inhibits the Growth of
Anaplastic Thyroid Cancer
Bradley A. Schiff, Andrea B. McMurphy, Samar A. Jasser,
Maher N.Younes, Dao Doan, Orhan G.Yigitbasi, Seungwon Kim,
Ge Zhou, Mahitosh Mandal, Benjamin N. Bekele,
F. Christopher Holsinger, Steven I. Sherman, Sai-Ching Yeung,
Adel K. El-Naggar, and Jeffrey N. Myers

The editors are publishing this note to alert readers to a concern about this article (1):
in Fig. 2B, a splice line is missing in the NPA 187 lane of the b-actin panel.

Reference
1. Schiff BA, McMurphy AB, Jasser SA, Younes MN, Doan D, Yigitbasi OG, et al. Epidermal growth
factor receptor (EGFR) is overexpressed in anaplastic thyroid cancer, and the EGFR inhibitor
gefitinib inhibits the growth of anaplastic thyroid cancer. Clin Cancer Res 2004;10:8594–602.

Published first August 1, 2019.


Clin Cancer Res 2019;25:4862
doi: 10.1158/1078-0432.CCR-19-2052
2019 American Association for Cancer Research.

4862 Clin Cancer Res; 25(15) August 1, 2019


Epidermal Growth Factor Receptor (EGFR) Is Overexpressed
in Anaplastic Thyroid Cancer, and the EGFR Inhibitor
Gefitinib Inhibits the Growth of Anaplastic Thyroid Cancer
Bradley A. Schiff, Andrea B. McMurphy, Samar A. Jasser, et al.

Clin Cancer Res 2004;10:8594-8602.

Updated version Access the most recent version of this article at:
http://clincancerres.aacrjournals.org/content/10/24/8594

Cited articles This article cites 28 articles, 5 of which you can access for free at:
http://clincancerres.aacrjournals.org/content/10/24/8594.full#ref-list-1

Citing articles This article has been cited by 16 HighWire-hosted articles. Access the articles at:
http://clincancerres.aacrjournals.org/content/10/24/8594.full#related-urls

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Subscriptions Department at pubs@aacr.org.

Permissions To request permission to re-use all or part of this article, use this link
http://clincancerres.aacrjournals.org/content/10/24/8594.
Click on "Request Permissions" which will take you to the Copyright Clearance Center's
(CCC)
Rightslink site.

Downloaded from clincancerres.aacrjournals.org on April 14, 2021. © 2004 American Association for Cancer
Research.

You might also like